Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Cell ; 184(8): 2103-2120.e31, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33740419

RESUMEN

During cell migration or differentiation, cell surface receptors are simultaneously exposed to different ligands. However, it is often unclear how these extracellular signals are integrated. Neogenin (NEO1) acts as an attractive guidance receptor when the Netrin-1 (NET1) ligand binds, but it mediates repulsion via repulsive guidance molecule (RGM) ligands. Here, we show that signal integration occurs through the formation of a ternary NEO1-NET1-RGM complex, which triggers reciprocal silencing of downstream signaling. Our NEO1-NET1-RGM structures reveal a "trimer-of-trimers" super-assembly, which exists in the cell membrane. Super-assembly formation results in inhibition of RGMA-NEO1-mediated growth cone collapse and RGMA- or NET1-NEO1-mediated neuron migration, by preventing formation of signaling-compatible RGM-NEO1 complexes and NET1-induced NEO1 ectodomain clustering. These results illustrate how simultaneous binding of ligands with opposing functions, to a single receptor, does not lead to competition for binding, but to formation of a super-complex that diminishes their functional outputs.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas Ligadas a GPI/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Oncogénicas/metabolismo , Animales , Moléculas de Adhesión Celular Neuronal/química , Movimiento Celular , Receptor DCC/deficiencia , Receptor DCC/genética , Proteínas Ligadas a GPI/química , Conos de Crecimiento/fisiología , Humanos , Ventrículos Laterales/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/química , Neuronas/citología , Neuronas/metabolismo , Proteínas Oncogénicas/química , Proteínas Oncogénicas/genética , Unión Proteica , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal
2.
J Neurol Neurosurg Psychiatry ; 94(8): 649-656, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36737245

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with limited treatment options and an incompletely understood pathophysiology. Although genomewide association studies (GWAS) have advanced our understanding of the disease, the precise manner in which risk polymorphisms contribute to disease pathogenesis remains unclear. Of relevance, GWAS have shown that a polymorphism (rs12608932) in the UNC13A gene is associated with risk for both ALS and frontotemporal dementia (FTD). Homozygosity for the C-allele at rs12608932 modifies the ALS phenotype, as these patients are more likely to have bulbar-onset disease, cognitive impairment and FTD at baseline as well as shorter survival. UNC13A is expressed in neuronal tissue and is involved in maintaining synaptic active zones, by enabling the priming and docking of synaptic vesicles. In the absence of functional TDP-43, risk variants in UNC13A lead to the inclusion of a cryptic exon in UNC13A messenger RNA, subsequently leading to nonsense mediated decay, with loss of functional protein. Depletion of UNC13A leads to impaired neurotransmission. Recent discoveries have identified UNC13A as a potential target for therapy development in ALS, with a confirmatory trial with lithium carbonate in UNC13A cases now underway and future approaches with antisense oligonucleotides currently under consideration. Considering UNC13A is a potent phenotypic modifier, it may also impact clinical trial outcomes. This present review describes the path from the initial discovery of UNC13A as a risk gene in ALS to the current therapeutic options being explored and how knowledge of its distinct phenotype needs to be taken into account in future trials.


Asunto(s)
Esclerosis Amiotrófica Lateral , Demencia Frontotemporal , Enfermedades Neurodegenerativas , Humanos , Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/complicaciones , Demencia Frontotemporal/patología , Enfermedades Neurodegenerativas/complicaciones , Proteínas del Tejido Nervioso/genética , Polimorfismo Genético
3.
Development ; 145(2)2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-29343636

RESUMEN

During development, precerebellar neurons migrate tangentially from the dorsal hindbrain to the floor plate. Their axons cross it but their cell bodies stop their ventral migration upon reaching the midline. It has previously been shown that Slit chemorepellents and their receptors, Robo1 and Robo2, might control the migration of precerebellar neurons in a repulsive manner. Here, we have used a conditional knockout strategy in mice to test this hypothesis. We show that the targeted inactivation of the expression of Robo1 and Robo2 receptors in precerebellar neurons does not perturb their migration and that they still stop at the midline. The selective ablation of the expression of all three Slit proteins in floor-plate cells has no effect on pontine neurons and only induces the migration of a small subset of inferior olivary neurons across the floor plate. Likewise, we show that the expression of Slit proteins in the facial nucleus is dispensable for pontine neuron migration. Together, these results show that Robo1 and Robo2 receptors act non-cell autonomously in migrating precerebellar neurons and that floor-plate signals, other than Slit proteins, must exist to prevent midline crossing.


Asunto(s)
Movimiento Celular/fisiología , Cerebelo/embriología , Glicoproteínas/fisiología , Proteínas del Tejido Nervioso/fisiología , Neuronas/fisiología , Receptores Inmunológicos/fisiología , Animales , Cerebelo/citología , Femenino , Glicoproteínas/deficiencia , Glicoproteínas/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Neurogénesis/fisiología , Embarazo , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética , Transducción de Señal , Proteínas Roundabout
4.
Development ; 145(2)2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-29343638

RESUMEN

During the development of the central nervous system (CNS), only motor axons project into peripheral nerves. Little is known about the cellular and molecular mechanisms that control the development of a boundary at the CNS surface and prevent CNS neuron emigration from the neural tube. It has previously been shown that a subset of spinal cord commissural axons abnormally invades sensory nerves in Ntn1 hypomorphic embryos and Dcc knockouts. However, whether netrin 1 also plays a similar role in the brain is unknown. In the hindbrain, precerebellar neurons migrate tangentially under the pial surface, and their ventral migration is guided by netrin 1. Here, we show that pontine neurons and inferior olivary neurons, two types of precerebellar neurons, are not confined to the CNS in Ntn1 and Dcc mutant mice, but that they invade the trigeminal, auditory and vagus nerves. Using a Ntn1 conditional knockout, we show that netrin 1, which is released at the pial surface by ventricular zone progenitors is responsible for the CNS confinement of precerebellar neurons. We propose, that netrin 1 distribution sculpts the CNS boundary by keeping CNS neurons in netrin 1-rich domains.


Asunto(s)
Sistema Nervioso Central/embriología , Sistema Nervioso Central/metabolismo , Netrina-1/metabolismo , Sistema Nervioso Periférico/embriología , Sistema Nervioso Periférico/metabolismo , Animales , Movimiento Celular/genética , Movimiento Celular/fisiología , Sistema Nervioso Central/citología , Receptor DCC/deficiencia , Receptor DCC/genética , Receptor DCC/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Netrina-1/deficiencia , Netrina-1/genética , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuronas/citología , Neuronas/metabolismo , Sistema Nervioso Periférico/citología , Embarazo
5.
Development ; 139(18): 3326-31, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22912413

RESUMEN

Gonadotropin-releasing hormone (GnRH) neurons are born in the nasal placode and migrate along olfactory and vomeronasal axons to reach the forebrain and settle in the hypothalamus, where they control reproduction. The molecular cues that guide their migration have not been fully identified, but are thought to control either cell movement directly or the patterning of their axonal substrates. Using genetically altered mouse models we show that the migration of GnRH neurons is directly modulated by Slit2 and Robo3, members of the axon guidance Slit ligand and Robo receptor families. Mice lacking Slit2 or Robo3 have a reduced number of GnRH neurons in the forebrain, but a normal complement of their supporting axons, pointing to a direct role for these molecules in GnRH neuron migration.


Asunto(s)
Movimiento Celular/fisiología , Hormona Liberadora de Gonadotropina/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuronas/metabolismo , Animales , Células COS , Movimiento Celular/genética , Chlorocebus aethiops , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas de la Membrana/genética , Ratones , Ratones Mutantes , Proteínas del Tejido Nervioso/genética , Receptores de Superficie Celular
6.
Circ Res ; 112(3): 465-75, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23255421

RESUMEN

RATIONALE: The Slit-Roundabout (Robo) signaling pathway has pleiotropic functions during Drosophila heart development. However, its role in mammalian heart development is largely unknown. OBJECTIVE: To analyze the role of Slit-Robo signaling in the formation of the pericardium and the systemic venous return in the murine heart. METHODS AND RESULTS: Expression of genes encoding Robo1 and Robo2 receptors and their ligands Slit2 and Slit3 was found in or around the systemic venous return and pericardium during development. Analysis of embryos lacking Robo1 revealed partial absence of the pericardium, whereas Robo1/2 double mutants additionally showed severely reduced sinus horn myocardium, hypoplastic caval veins, and a persistent left inferior caval vein. Mice lacking Slit3 recapitulated the defects in the myocardialization, alignment, and morphology of the caval veins. Ligand binding assays confirmed Slit3 as the preferred ligand for the Robo1 receptor, whereas Slit2 showed preference for Robo2. Sinus node development was mostly unaffected in all mutants. In addition, we show absence of cross-regulation with previously identified regulators Tbx18 and Wt1. We provide evidence that pericardial defects are created by abnormal localization of the caval veins combined with ectopic pericardial cavity formation. Local increase in neural crest cell death and impaired neural crest adhesive and migratory properties underlie the ectopic pericardium formation. CONCLUSIONS: A novel Slit-Robo signaling pathway is involved in the development of the pericardium, the sinus horn myocardium, and the alignment of the caval veins. Reduced Slit3 binding in the absence of Robo1, causing impaired cardiac neural crest survival, adhesion, and migration, underlies the pericardial defects.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Pericardio/metabolismo , Receptores Inmunológicos/metabolismo , Transducción de Señal , Venas Cavas/metabolismo , Animales , Apoptosis , Adhesión Celular , Movimiento Celular , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Morfogénesis , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Cresta Neural/anomalías , Cresta Neural/metabolismo , Pericardio/anomalías , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética , Nodo Sinoatrial/anomalías , Nodo Sinoatrial/metabolismo , Proteínas de Dominio T Box/metabolismo , Técnicas de Cultivo de Tejidos , Venas Cavas/anomalías , Proteínas WT1/metabolismo , Proteínas Roundabout
7.
Nat Commun ; 15(1): 4893, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38849340

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a debilitating motor neuron disease and lacks effective disease-modifying treatments. This study utilizes a comprehensive multiomic approach to investigate the early and sex-specific molecular mechanisms underlying ALS. By analyzing the prefrontal cortex of 51 patients with sporadic ALS and 50 control subjects, alongside four transgenic mouse models (C9orf72-, SOD1-, TDP-43-, and FUS-ALS), we have uncovered significant molecular alterations associated with the disease. Here, we show that males exhibit more pronounced changes in molecular pathways compared to females. Our integrated analysis of transcriptomes, (phospho)proteomes, and miRNAomes also identified distinct ALS subclusters in humans, characterized by variations in immune response, extracellular matrix composition, mitochondrial function, and RNA processing. The molecular signatures of human subclusters were reflected in specific mouse models. Our study highlighted the mitogen-activated protein kinase (MAPK) pathway as an early disease mechanism. We further demonstrate that trametinib, a MAPK inhibitor, has potential therapeutic benefits in vitro and in vivo, particularly in females, suggesting a direction for developing targeted ALS treatments.


Asunto(s)
Esclerosis Amiotrófica Lateral , Modelos Animales de Enfermedad , Sistema de Señalización de MAP Quinasas , Ratones Transgénicos , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Esclerosis Amiotrófica Lateral/metabolismo , Humanos , Femenino , Animales , Masculino , Ratones , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Piridonas/farmacología , Piridonas/uso terapéutico , Proteína FUS de Unión a ARN/metabolismo , Proteína FUS de Unión a ARN/genética , Corteza Prefrontal/metabolismo , Transcriptoma , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Persona de Mediana Edad , MicroARNs/genética , MicroARNs/metabolismo , Proteína C9orf72/genética , Proteína C9orf72/metabolismo , Caracteres Sexuales , Anciano , Factores Sexuales , Pirimidinonas
8.
Neuron ; 102(3): 512-514, 2019 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-31071282

RESUMEN

In this issue of Neuron, Bonanomi et al. (2019) investigate how navigating axons ignore irrelevant guidance signals. They show that the binding of p190RhoGAP to DCC suppresses inappropriate responses to Netrin-1, allowing motor axons to exit the embryonic spinal cord.


Asunto(s)
Orientación del Axón , Factores de Crecimiento Nervioso , Axones , Receptor DCC , Netrina-1 , Proteínas Supresoras de Tumor
9.
Cell Rep ; 9(4): 1191-201, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25456121

RESUMEN

Clearing techniques have been developed to transparentize mouse brains, thereby preserving 3D structure, but their complexity has limited their use. Here, we show that immunolabeling of axonal tracts followed by optical clearing with solvents (3DISCO) and light-sheet microscopy reveals brain connectivity in mouse embryos and postnatal brains. We show that the Robo3 receptor is selectively expressed by medial habenula axons forming the fasciculus retroflexus (FR) and analyzed the development of this commissural tract in mutants of the Slit/Robo and DCC/Netrin pathways. Netrin-1 and DCC are required to attract FR axons to the midline, but the two mutants exhibit specific and heterogeneous axon guidance defects. Moreover, floor-plate-specific deletion of Slit ligands with a conditional Slit2 allele perturbs not only midline crossing by FR axons but also their anteroposterior distribution. In conclusion, this method represents a unique and powerful imaging tool to study axonal connectivity in mutant mice.


Asunto(s)
Axones/metabolismo , Encéfalo/metabolismo , Imagenología Tridimensional/métodos , Coloración y Etiquetado , Animales , Biomarcadores/metabolismo , Receptor DCC , Embrión de Mamíferos/metabolismo , Proteínas de la Membrana/metabolismo , Ratones Noqueados , Mutación , Factores de Crecimiento Nervioso/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Netrina-1 , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/metabolismo , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/metabolismo
10.
Neuron ; 84(6): 1258-72, 2014 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-25433640

RESUMEN

Development of neuronal circuits is controlled by evolutionarily conserved axon guidance molecules, including Slits, the repulsive ligands for roundabout (Robo) receptors, and Netrin-1, which mediates attraction through the DCC receptor. We discovered that the Robo3 receptor fundamentally changed its mechanism of action during mammalian evolution. Unlike other Robo receptors, mammalian Robo3 is not a high-affinity receptor for Slits because of specific substitutions in the first immunoglobulin domain. Instead, Netrin-1 selectively triggers phosphorylation of mammalian Robo3 via Src kinases. Robo3 does not bind Netrin-1 directly but interacts with DCC. Netrin-1 fails to attract pontine neurons lacking Robo3, and attraction can be restored in Robo3(-/-) mice by expression of mammalian, but not nonmammalian, Robo3. We propose that Robo3 evolution was key to sculpting the mammalian brain by converting a receptor for Slit repulsion into one that both silences Slit repulsion and potentiates Netrin attraction.


Asunto(s)
Axones/metabolismo , Evolución Biológica , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Animales , Movimiento Celular , Receptor DCC , Glicoproteínas/metabolismo , Humanos , Ratones , Factores de Crecimiento Nervioso/metabolismo , Netrina-1 , Proteínas Supresoras de Tumor/metabolismo , Pez Cebra , Familia-src Quinasas/metabolismo
11.
J Biol Chem ; 283(47): 32792-801, 2008 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-18790729

RESUMEN

DM-GRASP, cell adhesion molecule of the immunoglobulin superfamily, has been shown to promote growth and navigation of axons. We here demonstrate that clustering of DM-GRASP in the plasma membrane induces its rapid internalization via dynamin- and clathrin-dependent endocytosis, which is controlled by phosphatidylinositol 3-kinase and mitogen-activated protein kinase ERK. The clustering of DM-GRASP activates ERK; the intensity and duration of ERK activation by DM-GRASP do not depend on rapid clathrin-mediated internalization of DM-GRASP. Moreover, the preference of retinal ganglion cell axons for DM-GRASP-coated micro-lanes requires clathrin-mediated endocytosis for the appropriate axonal turning reactions at substrate borders. Because the intracellular domain of DM-GRASP does not contain motifs for direct interactions with the endocytosis machinery, we performed a yeast two-hybrid screen to identify intracellular proteins mediating the uptake of DM-GRASP and isolated ubiquitin. Immunoprecipitation of DM-GRASP coexpressed with ubiquitin revealed that one or two ubiquitin(s) are attached to the intracellular domain of cell surface-resident DM-GRASP. Furthermore, elevated ubiquitination levels result in a decrease of cell surface-resident DM-GRASP as well as in the amount of total DM-GRASP. The endocytosis rate is not affected, but the delivery to multivesicular bodies is increased, indicating that DM-GRASP ubiquitination enhances its sorting into the degradation pathway. Together, our data show that ubiquitination and endocytosis of DM-GRASP in concert regulate its cell surface concentration, which is crucial for its function in axon navigation.


Asunto(s)
Axones/metabolismo , Endocitosis , Moléculas de Adhesión de Célula Nerviosa/química , Ubiquitina/metabolismo , Animales , Biotinilación , Adhesión Celular , Línea Celular , Membrana Celular/metabolismo , Pollos , Clatrina/química , Humanos , Microscopía Fluorescente , Modelos Biológicos , Retina/metabolismo , Ubiquitina/química
12.
Development ; 132(16): 3609-18, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16033798

RESUMEN

We investigated the role of the cell adhesion molecule NrCAM for axonal growth and pathfinding in the developing retina. Analysis of the distribution pattern of NrCAM in chick embryo retina sections and flat-mounts shows its presence during extension of retinal ganglion cell (RGC) axons; NrCAM is selectively present on RGC axons and is absent from the soma. Single cell cultures show an enrichment of NrCAM in the distal axon and growth cone. When offered as a substrate in addition to Laminin, NrCAM promotes RGC axon extension and the formation of growth cone protrusions. In substrate stripe assays, mimicking the NrCAM-displaying optic fibre layer and the Laminin-rich basal lamina, RGC axons preferentially grow on NrCAM lanes. The three-dimensional analysis of RGC growth cones in retina flat-mounts reveals that they are enlarged and form more protrusions extending away from the correct pathway under conditions of NrCAM-inhibition. Time-lapse analyses show that these growth cones pause longer to explore their environment, proceed for shorter time spans, and retract more often than under control conditions; in addition, they often deviate from the correct pathway towards the optic fissure. Inhibition of NrCAM in organ-cultured intact eyes causes RGC axons to misroute at the optic fissure; instead of diving into the optic nerve head, these axons cross onto the opposite side of the retina. Our results demonstrate a crucial role for NrCAM in the navigation of RGC axons in the developing retina towards the optic fissure, and also for pathfinding into the optic nerve.


Asunto(s)
Proteínas Aviares/metabolismo , Axones/metabolismo , Moléculas de Adhesión Celular/metabolismo , Movimiento Celular/fisiología , Conos de Crecimiento/metabolismo , Células Ganglionares de la Retina/metabolismo , Animales , Proteínas Aviares/genética , Moléculas de Adhesión Celular/genética , Células Cultivadas , Embrión de Pollo , Quiasma Óptico/embriología , Células Ganglionares de la Retina/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA